Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Oct 8.
Published in final edited form as: Cancer Lett. 2008 May 13;269(2):363–377. doi: 10.1016/j.canlet.2008.03.044

Multi-targeted Therapy of Cancer by Omega-3 Fatty Acids

Isabelle M Berquin 1,2, Iris J Edwards 2, Yong Q Chen 1,*
PMCID: PMC2572135  NIHMSID: NIHMS72462  PMID: 18479809

Abstract

Omega-3 (n-3) and omega-6 (n-6) polyunsaturated fatty acids (PUFAs) are essential fatty acids necessary for human health. Currently, the Western diet contains a disproportionally high amount of n-6 PUFAs and low amount of n-3 PUFAs, and the resulting high n-6/n-3 ratio is thought to contribute to cardiovascular disease, inflammation, and cancer. Studies in human populations have linked high consumption of fish or fish oil to reduced risk of colon, prostate and breast cancer, although other studies failed to find a significant association. Nonetheless, the available epidemiological evidence, combined with the demonstrated effects of n-3 PUFAs on cancer in animal and cell culture models, has motivated the development of clinical interventions using n-3 PUFAs in the prevention and treatment of cancer, as well as for nutritional support of cancer patients to reduce weight loss and modulate the immune system. In this review, we discuss the rationale for using long-chain n-3 PUFAs in cancer prevention and treatment and the challenges that such approaches pose in the design of clinical trials.

Introduction

Cardiovascular disease, cancer, obesity and diabetes collectively are responsible for more than 80% of the disease-related mortality in the US. Lipids play critical roles in all of these diseases, and the relative amounts and the types of dietary lipids consumed are believed to be of critical importance. Total fat intake and the ratio of n-6 to n-3 PUFAs in the Western diet have increased significantly since the Industrial Revolution (1, 2). Thus, today’s standard diet differs from the diet on which human beings have evolved, with profound implication on health. A preventive effect of n-3 PUFAs on cardiovascular disease, cancer as well as metabolic syndromes such as obesity and diabetes has been investigated extensively, and these studies were critically assessed in several Evidence Reports commissioned by the US Department of Health & Human Services (3, 4, 5, 6) and discussed in some recent reviews (7, 8, 9, 10, 11). An aspect that is not well understood to date is the importance of gene-environment and in particular gene-diet interaction in human health. Indeed, the amount and content of dietary fat can have a drastic impact on the health of animals with an identical genetic background as well as the health of genetically predisposed populations.

The purpose of this review is to highlight recent progress in our understanding of the molecular mechanisms of action of n-3 PUFAs, with an emphasis on their multi-targeted effects, and discuss clinical trials investigating the role of n-3 PUFAs in cancer prevention and treatment.

Dietary sources and metabolism of polyunsaturated fatty acids

Dietary sources of n-3 and n-6 fatty acids

Saturated and monounsaturated fatty acids can be synthesized de novo and obtained from the diet. However, mammals lack the desaturases necessary for synthesizing n-3 and n-6 PUFAs, therefore these essential fatty acids must be obtained from the diet. Terrestrial plants synthesize the first member of the n-6 series, linoleic acid (LA; 18:2n-6). LA is abundant in nearly all commonly available vegetable oils, including corn, sunflower, safflower and olive oil. Plants can also synthesize the first member of the n-3 PUFA series, alpha linolenic acid (α-LNA; 18:3n-3). Sources of this fatty acid include soybeans, walnuts, dark green leafy vegetables such as kale, spinach, broccoli and Brussels sprouts, and seeds or their oils such as flaxseed, mustard seed and rapeseed (canola); however, the majority of these oils are also rich in LA. Dietary long-chain n-3 PUFAs are found primarily in cold-water fish in forms of eicosapentaenoic acid (EPA; 20:5n-3) and docosahexaenoic acid (DHA; 22:6n-3). Fish ingest EPA and DHA from phytoplankton and zooplankton (12). It is important to note that the content of marine n-3 fatty acids varies greatly according to the species of fish, the total fat content of the fish and the geographical location of waters they inhabit (13). Even within a species, the n-3 fatty acids vary from Atlantic to Pacific Ocean. As a general rule, deep water fish such as mackerel, tuna and salmon from colder temperatures have the highest content of EPA and DHA. Fish farming may have a marked influence on fatty acid composition according to diets supplied to the fish (12).

Conversion of LA to AA and of α-LNA to EPA and DHA in animals and humans

While mammals cannot synthesize n-3 or -6 PUFAs de novo, mammalian cells can interconvert PUFAs within each series by elongation, desaturation and retroconversion (14). After ingestion, LA is metabolized by a series of alternating oxidative desaturation and elongation steps principally to arachidonic acid (AA; 20:4n-6). The main metabolic pathways for conversion of PUFAs are shown in Figure 1. The Δ6 pathway is responsible for conversion of LA to AA, and α-LNA to EPA, occurring primarily in the endoplasmic reticulum of liver cells. The Δ8 pathway, founded in plants, can also generate AA and EPA. Unlike α-LNA to EPA, the conversion from EPA to DHA is somewhat controversial. EPA may be converted to DHA through the Δ4 pathway (15). However, the existence of Δ4 fatty acid desaturation in the biosynthesis of DHA had been questioned for years, until a Δ4 fatty acid desaturase was characterized in vegetative organisms (16). Nevertheless, the existence of such desaturation for DHA synthesis in mammals is still unclear. Meanwhile, evidence suggests that the conversion to DHA occurs through the Sprecher pathway, namely elongation of docosapentaenoic acid (DPA; 22:5n-3) to tetracosahexaenoic acid (THA; 24:6n-3) and translocation to peroxisomes, a 2-carbon removal by β-oxidation, and translocation of DHA back to the endoplasmic reticulum (17).

Figure 1. Pathways for the synthesis of very long chain polyunsaturated fatty acids.

Figure 1

Linoleic acid (LA) and alpha-linolenic acid (α-LNA) are converted to arachidonic acid (AA) and eicosapentaenoic acid (EPA), respectively, through a sequential action of delta6 fatty acid desaturase (Δ6), elongase (elo) and delta5 fatty acid desaturase (Δ5). EPA may be elongated to docosapentaenoic acid (DPA) and then desaturated by delta4 fatty acid desaturase (Δ4) to docosahexaenoic acid (DHA). However, Sprecher et al. have demonstrated that EPA is likely elongated to DPA and to tetracosapentaenoic acid (TPA), desaturated to tetracosahexaenoic acid (THA) by Δ6 desaturase, translocated to peroxisomes, and β-oxidized (β-oxi) to docosahexaenoic acid (DHA).

The conversion of α-LNA to DHA in humans is very inefficient and since the same enzymes are involved in n-3 and n-6 pathways, the background diet may have a significant impact. Both α-LNA-feeding studies and stable isotope studies have consistently demonstrated that increased consumption of α-LNA does not result in increased DHA in plasma or cell lipids [reviewed in (18)]. In the first place most of the α-LNA is β-oxidized to provide energy and less than 10% of ingested α-LNA is converted to EPA. There is some evidence of gender differences in efficiency of the elongation-desaturation pathway suggesting that sex hormones may play a regulatory role, but this has not been widely studied (18). One study indicated that supplementing women with flaxseed oil (α-LNA) does not increase DHA (19). From kinetic analyses of fatty acid conversion, it was estimated that overall efficiency of conversion of α-LNA may be as low as 0.2% to EPA, 0.13% to DPA, and 0.05% to DHA (20). These data suggest that Δ-6 desaturase, the first enzyme in the bioconversion sequence, is the rate limiting step of the pathway. Recently, it was proposed that the limited conversion of α-LNA to DHA in HepG2 cells was due to a competition of α-LNA and tetracosapentaenoic acid (TPA; 24:5n-3) for Δ-6 desaturase (21). Alternatively, there is evidence for two different Δ-6 desaturase activities for these two steps (22). Moreover, studies by Park and Harris (23) have shown that human intake of 4 grams of EPA/day for a 4-month period did not result in a detectable increase in plasma DHA, indicating that even in the absence of high α-LNA, the EPA to DHA conversion is an inefficient process. Our own in vitro studies have shown that human breast and prostate cancer cell lines can effectively convert EPA to DPA but have little ability to proceed to any subsequent step (Edwards et al. unpublished). Despite the lack of understanding of many details in this pathway to guide nutritional formulations and recommendations, a consensus has emerged that the most effective way to increase plasma and tissue concentrations of a particular n-3 fatty acid is to feed that specific fatty acid (24). In addition, this also implies that the source and nature of n-3 PUFAs under investigation in any preclinical or clinical setting should be considered carefully in both the design and interpretation of the study.

Observational human studies linking PUFA intake and cancer risk

Epidemiological literature on the association of n-3 PUFAs and cancer, including correlational studies and migrational studies, suggests a protective effect of n-3 PUFAs and a promoting effect of n-6 PUFAs on cancer. However, the results of such studies are mixed, and many fail to demonstrate a statistically significant association between n-3 PUFAs and reduced cancer risk. Several recent reviews have been published on this topic (2, 11, 13, 25, 26, 27). Here, we will limit ourselves to a brief overview of observational studies that have been published since our last review (11) and discuss limitations inherent to the observational human studies. Recent studies supporting a protective role of n-3 PUFAs in cancer include case-control studies performed in Japan (28) and Scotland (29). In the Japanese study, there was a trend for an inverse relationship between the risk of colorectal cancer and n-3 PUFA consumption, but this association was only statistically significant for distal colon cancer. In the Scottish study, significant dose-dependant reductions in colon cancer risk were associated with increased intake of total n-3 PUFAs as well as EPA or DHA taken separately. An additional case-control study with measurement of blood fatty acid levels showed an inverse relationship between long-chain n-3 fatty acids and overall prostate cancer risk (30); interestingly, LA levels were also inversely correlated with prostate cancer risk, but LA metabolites were directly correlated with cancer risk. However, in a study of Jamaican men undergoing prostate biopsy for elevated PSA levels, a positive correlation was observed between the LA-to-DHA ratio measured in erythrocyte membranes and prostate tumor volume (31).

Several confounding factors could account for the inconsistent results obtained in epidemiological data investigating the association between n-3 PUFAs and cancer risk. First, there are wide variations in the amount and source of n-3 PUFAs consumed in each study. Population and ecological studies mainly rely on data from self reported dietary fatty acid intakes or from estimates based on national consumption, and these assessments correlate poorly with direct measurements of fatty acids in individual patient samples. In some studies, the actual intake in n-3 PUFAs may be too low for a protective effect, or the protective effect may be mitigated by contaminants or other dietary components. Second, the ratio of n-6 to n-3 may be more important than the absolute amount of n-3 PUFAs, as suggested by animal and human studies (2, 32, 33). Our results in prostate-specific Pten knockout mice indicated that a ratio of n-6:n-3 lower than 5 was effective in slowing cancer progression (33). Because individuals consume different amounts of n-6 PUFAs and may have genetic or epigenetic differences in enzymes responsible for lipid metabolism, the amount of n-3 intake has to be individualized in order to achieve an effective ratio. Third, it is possible that results will depend upon the type (α-LNA, EPA or DHA) and even the form (triglyceride or ester) of n-3 PUFAs used in various cancers. As mentioned earlier, it is still unclear whether different dietary n-3 PUFAs are equally effective in tumor suppression, and the conversion of α-LNA to EPA or DHA in humans is poor. Finally, the effects of n-3 PUFAs on cancer risk in an individual may be influenced by genetic factors, such as polymorphism in modifier genes. For instance, studies suggest a possible interaction between dietary intake of PUFAs and polymorphisms in cyclooxygenase (COX) and lipoxygenase (LOX) genes in determining cancer risk (34, 35, 36). As discussed in the next section, COX and LOX families of enzymes can metabolize both n-3 and -6 PUFAs to eicosanoids with different biological effects that could modulate cancer risk.

Molecular targets of PUFA

A number of biological effects that could contribute to cancer promotion by n-6 PUFAs and cancer suppression by n-3 PUFAs have been suggested (13, 37). These effects include alterations in the properties of cancer cells (proliferation, invasion, metastasis and apoptosis) as well as those of host cells (inflammation, immune response and angiogenesis). The molecular mechanisms which account for these biological effects are not completely understood. Some of the proposed molecular targets are discussed below.

Antagonism between n-3 and -6 PUFAs

The most widely studied effects of PUFAs are those that relate to eicosanoid biosynthesis and function. Eicosanoids are biologically active lipids with chain lengths of 20 carbon atoms which collectively modulate cell growth and differentiation, inflammation, immunity, platelet aggregation and angiogenesis. Dietary n-6 and n-3 PUFAs can be metabolized to prostaglandins (PG), thromboxanes (TX), hydroxyeicosatetraenoic acids (HETE) and leukotrienes (LT) by the enzymatic activity of COXs and LOXs. Generally speaking, eicosanoids derived from n-6 PUFAs have pro-inflammatory whereas those derived from n-3 precursors have anti-inflammatory effects (38). Likewise, eicosanoids derived from these two series have opposing effects in cancer cell growth (39, 40), invasion (41) and angiogenesis (42, 43). Besides eicosanoids, marine n-3 PUFAs can also be metabolized to resolvins and protectins (44, 45). These compounds possess potent anti-inflammatory and immunoregulatory actions (46). Mounting evidence suggests that inflammation may play a critical role in the development of human cancer (47, 48, 49). Therefore, one of the possible mechanisms for inhibition of tumors by n-3 PUFAs is by suppression of inflammation through resolvins.

In addition to the biological effects of eicosanoids and other metabolites, n-3 PUFAs are also thought to exert indirect effects by inhibiting n-6 series eicosanoid biosynthesis. n-3 PUFAs become incorporated into membrane phospholipids, where they partially replace AA and reduce the pool of available AA. They compete with n-6 PUFAs for desaturases, elongases, COXs and LOXs, so that the biosynthesis of n-6 series eicosanoids is reduced. For instance, the high binding affinity and poor substrate properties of EPA for COX-1 interferes with AA oxygenation by this enzyme (50, 51). Inhibition of tumor cell growth and invasion by n-3 PUFAs in a xenograft animal model was associated with decreased COX-2 and PGE2 levels (32). Thus n-3 PUFAs may act as a natural COX “inhibitor”.

Differential effects on signaling pathway, transcription and translation

A number of signaling pathways that are relevant to carcinogenesis and tumor progression are differentially affected by n-3 and -6 PUFAs. For instance, n-6 PUFA products were reported to upregulate and activate cellular signaling mediators including protein kinase C (42, 52), ras (53, 54), ERK 1/2 (55) and NF-κB (56) whereas n-3 PUFA products had the opposite effect. In Fat-1 transgenic mice, which convert endogenous n-6 to n-3 PUFAs, EPA and DHA activated the Jak2/Stat5 pathway in mammary cells, leading to cell differentiation (57). For the most part, signaling pathways converge in the nucleus to regulate transcription factor function, leading to alterations in gene expression. The pleiotropic nature of transcriptional changes induced by n-3 and -6 PUFAs was illustrated by studies where global gene expression patterns were determined by microarray analysis in vitro (58) or in vivo (59). Several hundreds of genes participating in a number of biological processes are differentially regulated by n-3 and -6 PUFAs, underlining the complexity of the transcriptional changes induced - directly or indirectly - by these fatty acids.

The exact molecular mechanism of these signaling and transcriptional effects is not clear. Changes in lipid composition of the plasma membrane may affect the membrane fluidity and the way growth factors, cytokines and hormones interact with their receptors, and the resulting signal transduction through secondary messengers. One site of particular interest within the plasma membrane is the lipid raft, a microdomain thought to play important roles in signaling (60). EPA and DHA were shown to alter the lipid raft composition in MDA-MB-231 breast cancer cells, leading to sustained phosphorylation of epidermal growth factor receptor and downstream signaling pathways, but paradoxically reducing cell growth (61).

A second type of mechanism through which n-3 fatty acids may alter cellular signaling is by acting directly as ligands for nuclear receptors, including peroxisome proliferators-activated receptors (PPARs) (62) or retinoid X receptor alpha (63). These nuclear transcription factors bind lipid ligands to regulate gene expression, thereby mediating biological functions ranging from lipid metabolism and homeostasis to cell differentiation and cell death. DHA was shown to induce cell apoptosis through activation of PPARγ (64) Edwards et al, unpublished). The levels of nuclear receptor expressed in cells may also be modulated by PUFAs (63, 65).

n-3 PUFAs may also regulate the translation machinery. EPA has been reported to affect intracellular homeostasis: EPA induced a Ca++ release from the intracellular Ca++ stores and simultaneously inhibited Ca++ influx through store-operated Ca++ channels in the plasma membrane, resulting in a depletion of the intracellular Ca++ stores. Such Ca++ depletion activated eIF2α kinase, inhibited translation initiation, and preferentially down-regulated oncogenes and G1 cyclins (66). It is unclear if other n-3 PUFAs, namely α-LNA and DHA, have similar effects.

Recent data suggested that Bcl-2 family proteins may play a key role in n-3 PUFA-induced cell death. We found that n-3 fatty acids reduced prostate tumor growth, slowed histopathological progression and increased survival of Pten knockout mice, whereas n-6 fatty acids had opposite effects. Tumors from mice on an n-3-enriched diet had lower proportions of phosphorylated Bad and higher apoptotic indexes compared to those on an n-6-rich diet. Knockdown of Bad eliminated n-3-induced cell death, and introduction of exogenous Bad restored the sensitivity to n-3 fatty acids (33). Furthermore, knockout of Bad abolished the tumor suppressive effect of n-3 fatty acids in vivo (Chen et al., unpublished). Our data suggest that modulation of prostate cancer development by PUFAs is mediated in part through Bad-dependent apoptosis.

Role of lipid peroxidation in PUFA-induced cytotoxicity

The ability of long-chain n-3 PUFAs to induce apoptosis in tumor cells has been attributed to the increased susceptibility of these cells to lipid peroxidation (62, 67). In support of this mechanism, the ability of fatty acid preparations to induce cell death was linked to their ability to increases the level of secondary products of lipid peroxidation. In addition, the tumor suppressive effect of n-3 PUFAs in human breast and colon cancer cell lines was blocked in the presence of antioxidants. As n-6 PUFAs are also susceptible to undergo auto-oxidation, it is possible that peroxidized products of n-3 and n-6 PUFAs have different biological properties.

Animal studies supporting a protective role of n-3 PUFAs on cancer development

The strongest data supporting a tumor protective role for n-3 PUFAs is provided by animal studies. Here, we will briefly outline some of the evidence obtained in animal models of prostate, breast and colon cancer.

Models of prostate cancer

Early studies showed that growth of human prostate cancer cell xenografts in nude mice was significantly inhibited by a fish oil diet (68, 69). More recently, a xenograft study using severe combined immunodeficiency mice demonstrated that tumor growth rate, final tumor volume, and serum PSA levels were all reduced by n-3 dietary supplementation (32). Decreased cyclooxygenase-2 (COX-2) and prostaglandin E2 were measured in tumor tissues from n-3-fed animals. In our own studies we have used prostate-specific Pten knock out mice that have the advantage of being an immune competent, orthotopic model with similarities in tumor development to human prostate cancer (70). In these animals an n-3 fatty acid-enriched diet reduced tumor growth, retarded histopathologic progression and extended lifespan, whereas n-6 fatty acid supplementation had the opposite effect (33). In addition, tumor growth was also retarded in this model by introduction of the fat-1 omega-3 desaturase which converts n-6 to n-3 fatty acids (33).

Models of breast cancer

A number of studies have shown n-3 fatty acid protection in rodent models of breast cancer. These include dietary supplementation of mouse transplantable tumors (71) and human cell xenograft models (72, 73) as well as chemically induced tumors in rats (74, 75, 76). A recent report showed that dietary DHA-induced reduction in mammary tumors in a rat model was accompanied by a 60% increase in BRCA1 tumor suppressor protein (77). Interestingly, n-3 fatty acid-enriched diets enhanced the efficiency of doxorubicin (78) and mitomycin C (79) in inhibiting tumor growth and strengthened the inhibitory effect of tamoxifen in estrogen-dependent xenografts (80). These studies point to a potential value of n-3 fatty acids as adjuvant to standard chemotherapy (81).

Models of colon cancer

As in prostate and breast cancer, efficacy of n-3 fatty acids protection in colon cancer is well supported by animal studies. These include mouse models with transplantable tumors (82, 83, 84, 85, 86), as well as rats with chemically-induced colon cancer (87, 88, 89, 90, 91). In addition to primary tumor growth suppression, effective inhibition of metastases was also seen with n-3 enriched diets (83, 92, 93). Several studies with colon cancer models have emphasized a critical role of the COX pathway in the protective effects of n-3 fatty acids (86, 87). However, tumor formation was inhibited in both COX-2 deficient as well as COX-2 over expressing colon cancer xenografts, suggesting the existence of a COX-2 independent pathway for n-3 fatty acid protection (85).

Although the tumor suppressive effect of marine n-3 PUFAs (EPA and DHA) is consistently observed, the effect of botanical n-3 PUFAs (α-LNA) is more controversial. Although a number of studies have indicated that flaxseed oil had anti-colon cancer activity in animal models (94, 95, 96), only EPA and DHA, but not α-LNA, reduced tumorigenesis in ApcMin/+ mice (97). Importantly, some evidence suggested that the level of α-LNA was positively associated with cancer risk in human prostate cancer patients (98, 99). This inconsistency, together with the fact that there is little conversion of α-LNA to EPA or DHA in humans, warrants further investigation.

Clinical trials using n-3 PUFAs for cancer prevention and treatment

The tantalizing epidemiological data obtained to date, combined with the demonstrated effects of n-3 PUFAs on cancer in animal and cell culture models, has motivated the development of clinical intervention trials using fish oil or n-3 PUFAs in the prevention and treatment of cancer, as well as for nutritional support of cancer patients to reduce weight loss and modulate the immune system. Table I summarizes ongoing or recently completed clinical trials listed in the ClinicalTrials.gov database (http://clinicaltrials.gov/) as of January 2008 with dietary interventions including n-3 PUFA supplements. The majority of these trials have not yet been published. Published trials include NCT00031707 (100), NCT 00003077 (101) and NCT00558155 (102). In addition, the methodology of trial NCT 00094562 and the issues affecting dietary studies design were discussed (103).

Table I.

Intervention trials with omega-3 PUFA for cancer prevention, treatment or patient support.

Trial no. Cancer type Trial type Title Outcomes to be measured Dates/Reference Phase
NCT00114296 Breast cancer Prev. Omega-3 Fatty Acids in Preventing Breast Cancer in Women at High Risk of Developing Breast Cancer Mammographic breast density, atypia in ductal lavage, hormones, lipid peroxidation 04/05-NS NS
NCT00432913 Colon cancer Prev. Effect of 2 Doses of EPA on Apoptosis and Cell Proliferation on Colon Mucosa Proliferation and apoptosis in colonic mucosa, measure of tissue EPA, safety and tolerability 10/06-05/08 II/III
NCT00510692 Colon cancer Prev. Chemoprevention Trial in Familial Adenomatous Polyposis Coli Using EPA Number/ size of rectal polyps, apoptosis and cell proliferation in the rectal mucosa, EPA and FA uptake, tolerability 11/06-03/08 II/III
NCT00475722 Colon cancer Prev. Healthy Eating for Colon Cancer Prevention FA, carotenoids, inflammation mediators 05/07-10/12 NS
NCT00398333 Colorectal cancer Supp. Study to Assess the Effectiveness of a Omega-3 Enriched Supplement on Chemotherapy Tolerance in Colon Cancer Patients Tolerance to chemotherapy (QoL), nutritional status 06/05-NS IV
NCT00488904 Colorectal cancer Supp. Omega-3 Fatty Acids and Postoperative Complications After Colorectal Surgery Reduction in infectious complications, hospitalization time, mortality 06/07-01/08 IV
NCT00145015 Colorectal cancer Prev. FishGastro Study: Fish Consumption and Gastro-Intestinal Health Apoptosis, cell prolif., lymphocyte infiltration, cytokines, prostaglandins, gene expression 12/04-01/08 NS
NCT00168987 Colorectal cancer, hepatocellular ca, cholangiocarcinoma Supp. Influence of an Oral Nutritional Supplement Rich in Omega-3 Fatty Acids on Functional State and Quality of Life in Malnourished Patients With Gastroenterological Tumors Muscle function, cognitive functions, QoL, tolerance of nutritional supplement, nutritional state 01/07-06/07 IV
NCT00558155 Gastric, pancreatic Supp. The Impact of Immunostimulating Nutrition on the Outcome of Surgery Surgical and nonsurgical complications, overall morbidity/mortality, hospital stay 06/01-12/05 Klek et al, 2005 IV
NCT00559156 Head and neck Supp. Arginine/Omega-3 Fatty Acids/Nucleotides Nutritional Supplement in Treating Patients With Stage III or Stage IV Head and Neck Cancer Undergoing Chemotherapy and Radiation Therapy Immunomodulatory effect 06/05-NS II
NCT00333099 Head and neck, esophageal Supp. INEC Study: Immuno-Modulating Enteral Nutrition in Cancer Anti-inflammation (mucositis),complications, nutritional and immune status, QoL, cost 05/06-04/09 IV
NCT00533078 Hematologic malignancies (AML) Supp. Lipid Use, Nutrition, and Colitis in Patients With Hematological Malignancies Anti-inflammation (colitis) 11/07-12/08 II
NCT00003077 Leukemia, Lymphoma, Multiple Myeloma, Plasma Cell Neoplasm Supp., treat. Omega-3 Fatty Acids in Treating Patients With Advanced Cancer Who Have Significant Weight Loss Weight loss prevention, n-3 PUFA maximum tolerated dose, antitumor response 10/95-NS Burns et al, 2004 I/II
NCT00455416 Lymphoma Treat. Dietary Intervention in Follicular Lymphoma Tumor cell proliferation, apoptosis, immune cell infiltrate, cytokines 04/07-12/09 II
NCT00292279 Multiple cancers Supp. The Impact of Fish Oil Emulsion on Clinical Outcome of Post-Operative Cancer Patients Infection, inflammation, hosp. days, cost 06/02-03/06 III
NCT00031707 Multiple cancers (excl. brain, breast, prostate, ovarian, endometrial) Supp. Comparison of Megestrol and/or Omega-3 Fatty Acid-Enriched Nutritional Supplement in Treating Patients With Cancer-Related Weight Loss and Lack of Appetite Weight, appetite, nausea, vomiting, QoL, toxicity, overall survival, IL-6 03/00-NS Jatoi et al, 2004 III
NCT00094562 Multiple cancers and other diseases associated with cachexia Supp. A Fish Oil Supplement Maintain Body Weight Patients With Disease-Related Weight Loss Body weight, lean body mass, QoL, anti-inflammatory, anticatabolic, anti-oxidant and anabolic actions 06/04-09/06 II
NCT00253643 Prostate cancer Prev. Fish Oil in Preventing Prostate Cancer in Patients With Prostatic Intraepithelial Neoplasia or Who Are at for Developing Prostate Cancer Proliferation, lipid metabolism: FASN, SREBP, apoptosis, bone loss 07/05-NS NS
NCT00049309 Prostate cancer Prev. Low-Fat Diet and/or Flaxseed in Preventing Prostate Cancer Proliferation, apoptosis, PSA and other biomarkers 01/03-05/06 II
NCT00402285 Prostate cancer Treat. Lycopene or Omega-3 Fatty Acid Nutritional Supplements in Treating Patients With Stage I or Stage II Prostate Cancer Gene expression pattern, correlation with diet, lack of progression 04/03-NS NS
NCT00458549 Prostate cancer Treat. Polyunsaturated Fatty Acids in Treating Patients With Prostate Cancer Undergoing Prostate Biopsy and/or Surgery eIF2α phosphorylation, relapse-free survival 07/06-NS NS
NCT00433797 Prostate cancer Treat. Dietary Intervention With Phytochemicals and Polyunsaturated Fatty Acids in Prostate Cancer Patients PSA, biomarkers of inflammation, antioxidant, oxidative stress, oxidative damage 03/07-03/09 I/II

Prev.: prevention, Supp.: support, Treat.: treatment, NS: not specified, QoL: quality of life.

Nutritional support of cancer patients with n-3 PUFA supplements

Several trials address the possibility that nutritional supplements containing n-3 PUFAs are beneficial to reduce cancer-associated weight loss (cachexia) for patients with advanced malignancies. Initial clinical trials suggested that n-3 PUFAs could stabilize weight loss or lead to weight gain in advanced cancer patients with cachexia (104, 105, 106, 107). The results of larger trials have been mixed, due in part to problems with tolerability of the dietary supplements (100, 101, 108, 109). A recent review of the literature failed to find sufficient evidence to support the use of oral EPA in treatment of cancer cachexia (110). However, when patients were able to consume high doses of the dietary supplement, weight stabilization or weight gains could be achieved (101, 109). This suggests that n-3 PUFAs may have a role in nutritional support of cancer patients if they can be administered in such a way that high doses can be achieved for prolonged periods of time with limited gastrointestinal side effects. Several additional trials listed in Table I are investigating the possible role of n-3 dietary supplements in improving nutritional status of patients with colorectal cancer, head-and-neck cancer, and other cancer types. Interestingly, synthetic COX-2 inhibitors such as celecoxib may also be beneficial for cachexic cancer patients, alone or in combination with n-3 PUFAs (111, 112).

Nutritional supplements enriched in n-3 PUFAs have also been tested in clinical trials for their ability to improve the outcome of other cancer treatments. A particular area of emphasis has been placed on immunomodulatory effects of n-3 PUFAs, owing to their ability to reduce infection and inflammation (102, 113, 114). Administration of n-3 PUFAs either before or after major abdominal surgery for cancer was shown to reduce inflammatory cytokines (113, 115) and improve liver and pancreas function (116). Patients receiving parenteral immunonutrition with a nutritional supplement containing glutamine or n-3 PUFAs had reduced incidence of infectious complications and improved immune function compared to patients who received a nutritional supplement without glutamine or n-3 PUFAs (102). Bougnoux et al. reported that dietary supplementation of breast cancer patients with DHA during anthracyclin chemotherapy had beneficial effects on time to progression, overall survival, and tolerance of side effects, particularly in a group of patients with high incorporation of DHA as measured in the plasma (117). Ongoing studies listed in Table I include several additional trials to examine the ability of n-3 supplements to reduce postoperative complications after surgery in patients with colorectal, gastric and pancreatic cancer, and on reducing inflammation in patients with head-and-neck, esophageal, hematologic malignancies and other cancer types. In addition, a phase IV trial is under way to assess the effectiveness of n-3 supplements in improving tolerance to chemotherapy in patients with colon cancer.

Cancer prevention and treatment with n-3 PUFA supplements

Some evidence suggests that COX inhibitors, which primarily block the metabolism of AA, are beneficial in the prevention of colon (118) and prostate (119) cancer. However, the cardiovascular toxicity of COX-2 inhibitors (120) has jeopardized the clinical utility of these drugs. Reducing the intake of n-6 PUFAs and increasing the proportion of n-3 in our diet is an attractive approach to reduce the production of proinflammatory eicosanoids and prevent cancer. Prevention trials using n-3 supplements to reduce colorectal cancer risk have used cell proliferation in the colonic or rectal mucosa as an intermediate marker of cancer risk. A randomized double-blind trial found that administration of fish oil for 30 days to individuals with high risk for developing colon cancer reduced rectal mucosa proliferation to a level found in low risk population, with a concomitant increase in n-3 fatty acid and decrease in arachidonic acid in the rectal mucosa (121). A double-blind, crossover trial where healthy individuals were given a controlled basal diet supplemented with either fish oil or corn oil for 4 weeks showed that rectal cell proliferation, ornithine decarboxylase activity and PGE2 release were significantly lower during the fish oil period than the corn oil period (122). However, when a similar trial was repeated in the context of a high-fat diet (50% of energy), no significant differences were found, suggesting that the n-6:n-3 ratio is crucial in determining the effects of fish oil on cancer risk (123). At least four additional colorectal cancer prevention trials are ongoing to further define the role of n-3 PUFAs in colorectal cell proliferation and/or apoptosis as well as inflammation (Table I).

A second cancer type with potential chemopreventive benefit for n-3 PUFAs is prostate cancer, due in part to the extended window of opportunity afforded by the relatively slow disease progression. Preliminary studies have shown that a 3-month intervention with a low-fat, fish oil-supplemented diet was able to increase the n-3:n-6 ratio in plasma and adipose tissue (124). Another pilot study suggested that a low-fat, flaxseed-supplemented diet decreased PSA and cholesterol levels as well as benign prostatic epithelial cell proliferation in biopsy specimen from patients that were schedule for a repeat prostate biopsy (125). A phase II randomized controlled trial by the same group (NCT00049309) was completed recently and, to our knowledge, has not yet been published. Another clinical trial of prostate cancer prevention by n-3 PUFAs (NCT00049309) chose to use fish oil in patients with prostatic intraepithelial neoplasia or who are otherwise at risk for prostate cancer. In the latter study, still in progress, the measured outcomes include cell proliferation and apoptosis as well as phospholipid membrane composition, fatty acid synthase and sterol regulatory element binding protein expression. In addition to the prevention trials cited above, three prostate cancer trials listed in Table I include n-3 PUFA supplements as part of the treatment protocol for patients with early adenocarcinoma.

Dietary interventions with n-3 PUFAs have been assessed for prevention and/or treatment of other cancer types, including breast, skin, and lymphoma. An ongoing breast cancer prevention trial (NCT00114296) investigates the effect of n-3 PUFAs on mammographic breast density, breast epithelial cell atypia in ductal lavage samples, hormones and lipid peroxidation of women with family history of breast cancer or BRCA1/BRCA2 mutations. A possible beneficial role of n-3 PUFAs in skin cancer prevention was found in a double-blind randomized study of healthy subjects who took EPA for 3 months (126). The skin EPA content increased 8-fold from baseline, and sunburn sensitivity was reduced with a concomitant decrease in UVR-induced skin p53 expression. An ongoing Phase II clinical trial (NCT 00455416) is investigating the role of a dietary intervention including n-3 PUFAs, selenium, and extracts from garlic, pomegranate, grapes and green tea on follicular lymphoma proliferation, apoptosis, and immune function.

Conclusion

In recent history, the typical dietary composition in PUFAs has changed drastically, tipping the balance towards n-6 PUFAs, with negative consequences to human health. Restoring a healthier balance of n-3 to n-6 PUFAs is an attractive approach for cancer chemoprevention. Indeed, unlike many other chemopreventive agents, n-3 PUFAs are essential nutrients with additional beneficial effects on cardiovascular and inflammatory diseases. Experiments in animal models and tissue culture overwhelmingly support a protective effect of n-3 PUFAs against colon, prostate and breast cancer, whereas human association studies have been less consistent. Perhaps one reason for this difference is that diet, environment and genetic background can be controlled much easier in animal models than in human populations. Published epidemiological studies differ substantially in the source of n-3 PUFAs consumed (fish oils, seed oils, purified PUFAs), and few have confirmed the amounts of n-3 PUFAs available in blood or tissue samples by direct measurements. Because the conversion of α-LNA to EPA or DHA in humans is poor, results are likely to depend on the type (α-LNA, EPA or DHA) and even the form (triglyceride or ester) of n-3 PUFAs consumed, as well as the ratio of n-6 to n-3 PUFAs achieved in individuals, which will be influenced by dietary as well as genetic factors. Therefore, clinical trials with dietary interventions aiming to prevent or treat cancer need to be designed carefully to overcome these limitations and provide an optimal ratio of n-3 to n-6 PUFAs.

Acknowledgments

Our work is supported in part by NIH R01CA114017 (IMB), R01CA115958 (IJE), and R01CA107668 (YQC), P01CA106742 (YQC).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Simopoulos AP. Essential fatty acids in health and chronic disease. Am J Clin Nutr. 1999;70:560S–569S. doi: 10.1093/ajcn/70.3.560s. [DOI] [PubMed] [Google Scholar]
  • 2.Simopoulos AP. The importance of the ratio of omega-6/omega-3 essential fatty acids. Biomed Pharmacother. 2002;56:365–379. doi: 10.1016/s0753-3322(02)00253-6. [DOI] [PubMed] [Google Scholar]
  • 3.Wang C, Chung M, Balk E, Kupelnick B, DeVine D, Lawrence A, Lichtenstein A, Lau J. Effects of Omega-3 Fatty Acids on Cardiovascular Disease. U.S. Department of Health and Human Services Evidence Report/Technology Assessment. 2004;94 http://www.ahrq.gov/downloads/pub/evidence/pdf/o3cardio/o3cardio.pdf. [PMC free article] [PubMed]
  • 4.MacLean CH, Issa A, Khanna P, Lim Y, Mojica WA, Newberry SJ, Morton SC, Suttorp M, Tu W, Hilton LG, et al. Effects of Omega-3 Fatty Acids on Cancer. U.S. Department of Health and Human Services Evidence Report/Technology Assessment. 2005;113 http://www.ahrq.gov/downloads/pub/evidence/pdf/o3cancer/o3cancer.pdf. [PMC free article] [PubMed]
  • 5.Shekelle PG, Morton SC, MacLean CH, Garland RH, Tu W, Jungvig LK, Mojica WA, Pencharz J, Grossman J, Khanna P, et al. Effects of Omega-3 Fatty Acids on Lipids and Glycemic Control in Type II Diabetes and the Metabolic Syndrome and on Inflammatory Bowel Disease, Rheumatoid Arthritis, Renal Disease, Systemic Lupus Erythematosus, and Osteoporosis. U.S. Department of Health and Human Services Evidence Report/Technology Assessment. 2004;89 http://www.ahrq.gov/downloads/pub/evidence/pdf/o3lipid/o3lipid.pdf. [PMC free article] [PubMed]
  • 6.Balk E, Chung M, Lichtenstein A, Chew P, Kupelnick B, Lawrence A, DeVine D, Lau J. Effects of Omega-3 Fatty Acids on Cardiovascular Risk Factors and Intermediate Markers of Cardiovascular Disease. U.S. Department of Health and Human Services Evidence Report/Technology Assessment. 2004;93 http://www.ahrq.gov/downloads/pub/evidence/pdf/o3cardrisk/o3cardrisk.pdf. [PMC free article] [PubMed]
  • 7.Tziomalos K, Athyros VG, Mikhailidis DP. Fish oils and vascular disease prevention: an update. Curr Med Chem. 2007;14:2622–2628. doi: 10.2174/092986707782023307. [DOI] [PubMed] [Google Scholar]
  • 8.Jacobson TA. Beyond lipids: the role of omega-3 fatty acids from fish oil in the prevention of coronary heart disease. Curr Atheroscler Rep. 2007;9:145–153. doi: 10.1007/s11883-007-0011-x. [DOI] [PubMed] [Google Scholar]
  • 9.Carpentier YA, Portois L, Malaisse WJ. n-3 fatty acids and the metabolic syndrome. Am J Clin Nutr. 2006;83:1499S–1504S. doi: 10.1093/ajcn/83.6.1499S. [DOI] [PubMed] [Google Scholar]
  • 10.Barre DE. The role of consumption of alpha-linolenic, eicosapentaenoic and docosahexaenoic acids in human metabolic syndrome and type 2 diabetes--a mini-review. J Oleo Sci. 2007;56:319–325. doi: 10.5650/jos.56.319. [DOI] [PubMed] [Google Scholar]
  • 11.Chen YQ, Edwards IJ, Kridel SJ, Thornburg T, Berquin IM. Dietary fat-gene interactions in cancer. Cancer Metastasis Rev. 2007;26:535–551. doi: 10.1007/s10555-007-9075-x. [DOI] [PubMed] [Google Scholar]
  • 12.Sargent JR. Fish oils and human diet. Br J Nutr. 1997;78 Suppl 1:S5–S13. doi: 10.1079/bjn19970131. [DOI] [PubMed] [Google Scholar]
  • 13.Larsson SC, Kumlin M, Ingelman-Sundberg M, Wolk A. Dietary long-chain n-3 fatty acids for the prevention of cancer: a review of potential mechanisms. Am J Clin Nutr. 2004;79:935–945. doi: 10.1093/ajcn/79.6.935. [DOI] [PubMed] [Google Scholar]
  • 14.Crawford MA. The role of dietary fatty acids in biology: their place in the evolution of the human brain. Nutr Rev. 1992;50:3–11. doi: 10.1111/j.1753-4887.1992.tb01283.x. [DOI] [PubMed] [Google Scholar]
  • 15.Horrobin DF. Nutritional and medical importance of gamma-linolenic acid. Prog Lipid Res. 1992;31:163–194. doi: 10.1016/0163-7827(92)90008-7. [DOI] [PubMed] [Google Scholar]
  • 16.Qiu X, Hong H, MacKenzie SL. Identification of a Delta 4 fatty acid desaturase from Thraustochytrium sp. involved in the biosynthesis of docosahexanoic acid by heterologous expression in Saccharomyces cerevisiae and Brassica juncea. J Biol Chem. 2001;276:31561–31566. doi: 10.1074/jbc.M102971200. [DOI] [PubMed] [Google Scholar]
  • 17.Sprecher H. Metabolism of highly unsaturated n-3 and n-6 fatty acids. Biochim Biophys Acta. 2000;1486:219–231. doi: 10.1016/s1388-1981(00)00077-9. [DOI] [PubMed] [Google Scholar]
  • 18.Williams CM, Burdge G. Long-chain n-3 PUFA: plant v. marine sources. Proc Nutr Soc. 2006;65:42–50. doi: 10.1079/pns2005473. [DOI] [PubMed] [Google Scholar]
  • 19.Francois CA, Connor SL, Bolewicz LC, Connor WE. Supplementing lactating women with flaxseed oil does not increase docosahexaenoic acid in their milk. Am J Clin Nutr. 2003;77:226–233. doi: 10.1093/ajcn/77.1.226. [DOI] [PubMed] [Google Scholar]
  • 20.Pawlosky RJ, Hibbeln JR, Novotny JA, Salem N., Jr Physiological compartmental analysis of alpha-linolenic acid metabolism in adult humans. J Lipid Res. 2001;42:1257–1265. [PubMed] [Google Scholar]
  • 21.Portolesi R, Powell BC, Gibson RA. Competition between 24:5n-3 and ALA for Delta 6 desaturase may limit the accumulation of DHA in HepG2 cell membranes. J Lipid Res. 2007;48:1592–1598. doi: 10.1194/jlr.M700081-JLR200. [DOI] [PubMed] [Google Scholar]
  • 22.Marzo I, Alava MA, Pineiro A, Naval J. Biosynthesis of docosahexaenoic acid in human cells: evidence that two different delta 6-desaturase activities may exist. Biochim Biophys Acta. 1996;1301:263–272. doi: 10.1016/0005-2760(96)00051-3. [DOI] [PubMed] [Google Scholar]
  • 23.Park Y, Harris WS. Omega-3 fatty acid supplementation accelerates chylomicron triglyceride clearance. J Lipid Res. 2003;44:455–463. doi: 10.1194/jlr.M200282-JLR200. [DOI] [PubMed] [Google Scholar]
  • 24.Arterburn LM, Hall EB, Oken H. Distribution, interconversion, and dose response of n-3 fatty acids in humans. Am J Clin Nutr. 2006;83:1467S–1476S. doi: 10.1093/ajcn/83.6.1467S. [DOI] [PubMed] [Google Scholar]
  • 25.Terry PD, Rohan TE, Wolk A. Intakes of fish and marine fatty acids and the risks of cancers of the breast and prostate and of other hormone-related cancers: a review of the epidemiologic evidence. Am J Clin Nutr. 2003;77:532–543. doi: 10.1093/ajcn/77.3.532. [DOI] [PubMed] [Google Scholar]
  • 26.MacLean CH, Newberry SJ, Mojica WA, Khanna P, Issa AM, Suttorp MJ, Lim YW, Traina SB, Hilton L, Garland R, et al. Effects of omega-3 fatty acids on cancer risk: a systematic review. Jama. 2006;295:403–415. doi: 10.1001/jama.295.4.403. [DOI] [PubMed] [Google Scholar]
  • 27.Chen YQ, Berquin IM, Daniel LW, Edwards IJ, O'Flaherty JT, Thomas MJ, Tooze JA, Wykle B. Omega-3 fatty acids and cancer risk. Jama. 2006;296:282. doi: 10.1001/jama.296.3.282-a. author reply. [DOI] [PubMed] [Google Scholar]
  • 28.Kimura Y, Kono S, Toyomura K, Nagano J, Mizoue T, Moore MA, Mibu R, Tanaka M, Kakeji Y, Maehara Y, et al. Meat, fish and fat intake in relation to subsite-specific risk of colorectal cancer: The Fukuoka Colorectal Cancer Study. Cancer Sci. 2007;98:590–597. doi: 10.1111/j.1349-7006.2007.00425.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Theodoratou E, McNeill G, Cetnarskyj R, Farrington SM, Tenesa A, Barnetson R, Porteous M, Dunlop M, Campbell H. Dietary fatty acids and colorectal cancer: a case-control study. Am J Epidemiol. 2007;166:181–195. doi: 10.1093/aje/kwm063. [DOI] [PubMed] [Google Scholar]
  • 30.Chavarro JE, Stampfer MJ, Li H, Campos H, Kurth T, Ma J. A prospective study of polyunsaturated fatty acid levels in blood and prostate cancer risk. Cancer Epidemiol Biomarkers Prev. 2007;16:1364–1370. doi: 10.1158/1055-9965.EPI-06-1033. [DOI] [PubMed] [Google Scholar]
  • 31.Ritch CR, Wan RL, Stephens LB, Taxy JB, Huo D, Gong EM, Zagaja GP, Brendler CB. Dietary fatty acids correlate with prostate cancer biopsy grade and volume in Jamaican men. J Urol. 2007;177:97–101. doi: 10.1016/j.juro.2006.08.105. discussion 101. [DOI] [PubMed] [Google Scholar]
  • 32.Kobayashi N, Barnard RJ, Henning SM, Elashoff D, Reddy ST, Cohen P, Leung P, Hong-Gonzalez J, Freedland SJ, Said J, et al. Effect of altering dietary omega-6/omega-3 fatty acid ratios on prostate cancer membrane composition, cyclooxygenase-2, and prostaglandin E2. Clin Cancer Res. 2006;12:4662–4670. doi: 10.1158/1078-0432.CCR-06-0459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Berquin IM, Min Y, Wu R, Wu J, Perry D, Cline JM, Thomas MJ, Thornburg T, Kulik G, Smith A, et al. Modulation of prostate cancer genetic risk by omega-3 and omega-6 fatty acids. J Clin Invest. 2007;117:1866–1875. doi: 10.1172/JCI31494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Koh WP, Yuan JM, van den Berg D, Lee HP, Yu MC. Interaction between cyclooxygenase-2 gene polymorphism and dietary n-6 polyunsaturated fatty acids on colon cancer risk: the Singapore Chinese Health Study. Br J Cancer. 2004;90:1760–1764. doi: 10.1038/sj.bjc.6601797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Siezen CL, van Leeuwen AI, Kram NR, Luken ME, van Kranen HJ, Kampman E. Colorectal adenoma risk is modified by the interplay between polymorphisms in arachidonic acid pathway genes and fish consumption. Carcinogenesis. 2005;26:449–457. doi: 10.1093/carcin/bgh336. [DOI] [PubMed] [Google Scholar]
  • 36.Hedelin M, Chang ET, Wiklund F, Bellocco R, Klint A, Adolfsson J, Shahedi K, Xu J, Adami HO, Gronberg H, et al. Association of frequent consumption of fatty fish with prostate cancer risk is modified by COX-2 polymorphism. Int J Cancer. 2007;120:398–405. doi: 10.1002/ijc.22319. [DOI] [PubMed] [Google Scholar]
  • 37.Chapkin RS, McMurray DN, Lupton JR. Colon cancer, fatty acids and anti-inflammatory compounds. Curr Opin Gastroenterol. 2007;23:48–54. doi: 10.1097/MOG.0b013e32801145d7. [DOI] [PubMed] [Google Scholar]
  • 38.Calder PC, Yaqoob P, Thies F, Wallace FA, Miles EA. Fatty acids and lymphocyte functions. Br J Nutr. 2002;87 Suppl 1:S31–S48. doi: 10.1079/bjn2001455. [DOI] [PubMed] [Google Scholar]
  • 39.Abou-el-Ela SH, Prasse KW, Farrell RL, Carroll RW, Wade AE, Bunce OR. Effects of D,L-2-difluoromethylornithine and indomethacin on mammary tumor promotion in rats fed high n-3 and/or n-6 fat diets. Cancer Res. 1989;49:1434–1440. [PubMed] [Google Scholar]
  • 40.Rose DP, Connolly JM. Effects of fatty acids and inhibitors of eicosanoid synthesis on the growth of a human breast cancer cell line in culture. Cancer Res. 1990;50:7139–7144. [PubMed] [Google Scholar]
  • 41.Brown MD, Hart CA, Gazi E, Bagley S, Clarke NW. Promotion of prostatic metastatic migration towards human bone marrow stoma by Omega 6 and its inhibition by Omega 3 PUFAs. Br J Cancer. 2006;94:842–853. doi: 10.1038/sj.bjc.6603030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.McCarty MF. Fish oil may impede tumour angiogenesis and invasiveness by downregulating protein kinase C and modulating eicosanoid production. Med Hypotheses. 1996;46:107–115. doi: 10.1016/s0306-9877(96)90009-2. [DOI] [PubMed] [Google Scholar]
  • 43.Rose DP, Connolly JM. Regulation of tumor angiogenesis by dietary fatty acids and eicosanoids. Nutr Cancer. 2000;37:119–127. doi: 10.1207/S15327914NC372_1. [DOI] [PubMed] [Google Scholar]
  • 44.Serhan CN, Hong S, Gronert K, Colgan SP, Devchand PR, Mirick G, Moussignac RL. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med. 2002;196:1025–1037. doi: 10.1084/jem.20020760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Serhan CN, Arita M, Hong S, Gotlinger K. Resolvins, docosatrienes, and neuroprotectins, novel omega-3-derived mediators, and their endogenous aspirin-triggered epimers. Lipids. 2004;39:1125–1132. doi: 10.1007/s11745-004-1339-7. [DOI] [PubMed] [Google Scholar]
  • 46.Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol. 2005;6:1191–1197. doi: 10.1038/ni1276. [DOI] [PubMed] [Google Scholar]
  • 47.Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–867. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Karin M. Nuclear factor-kappaB in cancer development and progression. Nature. 2006;441:431–436. doi: 10.1038/nature04870. [DOI] [PubMed] [Google Scholar]
  • 49.Clevers H. At the crossroads of inflammation and cancer. Cell. 2004;118:671–674. doi: 10.1016/j.cell.2004.09.005. [DOI] [PubMed] [Google Scholar]
  • 50.Needleman P, Raz A, Minkes MS, Ferrendelli JA, Sprecher H. Triene prostaglandins: prostacyclin and thromboxane biosynthesis and unique biological properties. Proc Natl Acad Sci U S A. 1979;76:944–948. doi: 10.1073/pnas.76.2.944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wada M, DeLong CJ, Hong YH, Rieke CJ, Song I, Sidhu RS, Yuan C, Warnock M, Schmaier AH, Yokoyama C, et al. Enzymes and receptors of prostaglandin pathways with arachidonic acid-derived versus eicosapentaenoic acid-derived substrates and products. J Biol Chem. 2007;282:22254–22266. doi: 10.1074/jbc.M703169200. [DOI] [PubMed] [Google Scholar]
  • 52.Reddy BS, Simi B, Patel N, Aliaga C, Rao CV. Effect of amount and types of dietary fat on intestinal bacterial 7 alpha-dehydroxylase and phosphatidylinositol-specific phospholipase C and colonic mucosal diacylglycerol kinase and PKC activities during stages of colon tumor promotion. Cancer Res. 1996;56:2314–2320. [PubMed] [Google Scholar]
  • 53.Singh J, Hamid R, Reddy BS. Dietary fat and colon cancer: modulating effect of types and amount of dietary fat on ras-p21 function during promotion and progression stages of colon cancer. Cancer Res. 1997;57:253–258. [PubMed] [Google Scholar]
  • 54.Collett ED, Davidson LA, Fan YY, Lupton JR, Chapkin RS. n-6 and n-3 polyunsaturated fatty acids differentially modulate oncogenic Ras activation in colonocytes. Am J Physiol Cell Physiol. 2001;280:C1066–C1075. doi: 10.1152/ajpcell.2001.280.5.C1066. [DOI] [PubMed] [Google Scholar]
  • 55.Sauer LA, Blask DE, Dauchy RT. Dietary factors and growth and metabolism in experimental tumors. J Nutr Biochem. 2007;18:637–649. doi: 10.1016/j.jnutbio.2006.12.009. [DOI] [PubMed] [Google Scholar]
  • 56.Novak TE, Babcock TA, Jho DH, Helton WS, Espat NJ. NF-kappa B inhibition by omega -3 fatty acids modulates LPS-stimulated macrophage TNF-alpha transcription. Am J Physiol Lung Cell Mol Physiol. 2003;284:L84–L89. doi: 10.1152/ajplung.00077.2002. [DOI] [PubMed] [Google Scholar]
  • 57.Liu YE, Pu W, Wang J, Kang JX, Shi YE. Activation of Stat5 and induction of a pregnancy-like mammary gland differentiation by eicosapentaenoic and docosapentaenoic omega-3 fatty acids. Febs J. 2007;274:3351–3362. doi: 10.1111/j.1742-4658.2007.05869.x. [DOI] [PubMed] [Google Scholar]
  • 58.Edwards IJ, Berquin IM, Sun H, O'Flaherty JT, Daniel LW, Thomas MJ, Rudel LL, Wykle RL, Chen YQ. Differential effects of delivery of omega-3 fatty acids to human cancer cells by low-density lipoproteins versus albumin. Clin Cancer Res. 2004;10:8275–8283. doi: 10.1158/1078-0432.CCR-04-1357. [DOI] [PubMed] [Google Scholar]
  • 59.Berger A, Roberts MA, Hoff B. How dietary arachidonic- and docosahexaenoic-acid rich oils differentially affect the murine hepatic transcriptome. Lipids Health Dis. 2006;5:10. doi: 10.1186/1476-511X-5-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wassall SR, Brzustowicz MR, Shaikh SR, Cherezov V, Caffrey M, Stillwell W. Order from disorder, corralling cholesterol with chaotic lipids. The role of polyunsaturated lipids in membrane raft formation. Chem Phys Lipids. 2004;132:79–88. doi: 10.1016/j.chemphyslip.2004.09.007. [DOI] [PubMed] [Google Scholar]
  • 61.Schley PD, Brindley DN, Field CJ. (n-3) PUFA alter raft lipid composition and decrease epidermal growth factor receptor levels in lipid rafts of human breast cancer cells. J Nutr. 2007;137:548–553. doi: 10.1093/jn/137.3.548. [DOI] [PubMed] [Google Scholar]
  • 62.Stoll BA. N-3 fatty acids and lipid peroxidation in breast cancer inhibition. Br J Nutr. 2002;87:193–198. doi: 10.1079/BJNBJN2001512. [DOI] [PubMed] [Google Scholar]
  • 63.Fan YY, Spencer TE, Wang N, Moyer MP, Chapkin RS. Chemopreventive n-3 fatty acids activate RXRalpha in colonocytes. Carcinogenesis. 2003;24:1541–1548. doi: 10.1093/carcin/bgg110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zand H, Rhimipour A, Bakhshayesh M, Shafiee M, Nour Mohammadi I, Salimi S. Involvement of PPAR-gamma and p53 in DHA-induced apoptosis in Reh cells. Mol Cell Biochem. 2007;304:71–77. doi: 10.1007/s11010-007-9487-5. [DOI] [PubMed] [Google Scholar]
  • 65.Chambrier C, Bastard JP, Rieusset J, Chevillotte E, Bonnefont-Rousselot D, Therond P, Hainque B, Riou JP, Laville M, Vidal H. Eicosapentaenoic acid induces mRNA expression of peroxisome proliferator-activated receptor gamma. Obes Res. 2002;10:518–525. doi: 10.1038/oby.2002.70. [DOI] [PubMed] [Google Scholar]
  • 66.Aktas H, Halperin JA. Translational regulation of gene expression by omega-3 fatty acids. J Nutr. 2004;134:2487S–2491S. doi: 10.1093/jn/134.9.2487S. [DOI] [PubMed] [Google Scholar]
  • 67.Welsch CW. Review of the effects of dietary fat on experimental mammary gland tumorigenesis: role of lipid peroxidation. Free Radic Biol Med. 1995;18:757–773. doi: 10.1016/0891-5849(94)00187-o. [DOI] [PubMed] [Google Scholar]
  • 68.Karmali RA, Reichel P, Cohen LA, Terano T, Hirai A, Tamura Y, Yoshida S. The effects of dietary omega-3 fatty acids on the DU-145 transplantable human prostatic tumor. Anticancer Res. 1987;7:1173–1179. [PubMed] [Google Scholar]
  • 69.Rose DP, Cohen LA. Effects of dietary menhaden oil and retinyl acetate on the growth of DU 145 human prostatic adenocarcinoma cells transplanted into athymic nude mice. Carcinogenesis. 1988;9:603–605. doi: 10.1093/carcin/9.4.603. [DOI] [PubMed] [Google Scholar]
  • 70.Wang S, Gao J, Lei Q, Rozengurt N, Pritchard C, Jiao J, Thomas GV, Li G, Roy-Burman P, Nelson PS, et al. Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer Cell. 2003;4:209–221. doi: 10.1016/s1535-6108(03)00215-0. [DOI] [PubMed] [Google Scholar]
  • 71.Gabor H, Hillyard LA, Abraham S. Effect of dietary fat on growth kinetics of transplantable mammary adenocarcinoma in BALB/c mice. J Natl Cancer Inst. 1985;74:1299–1305. [PubMed] [Google Scholar]
  • 72.Rose DP, Connolly JM. Effects of dietary omega-3 fatty acids on human breast cancer growth and metastases in nude mice. J Natl Cancer Inst. 1993;85:1743–1747. doi: 10.1093/jnci/85.21.1743. [DOI] [PubMed] [Google Scholar]
  • 73.Rose DP, Connolly JM, Rayburn J, Coleman M. Influence of diets containing eicosapentaenoic or docosahexaenoic acid on growth and metastasis of breast cancer cells in nude mice. J Natl Cancer Inst. 1995;87:587–592. doi: 10.1093/jnci/87.8.587. [DOI] [PubMed] [Google Scholar]
  • 74.Jurkowski JJ, Cave WT., Jr Dietary effects of menhaden oil on the growth and membrane lipid composition of rat mammary tumors. J Natl Cancer Inst. 1985;74:1145–1150. [PubMed] [Google Scholar]
  • 75.Braden LM, Carroll KK. Dietary polyunsaturated fat in relation to mammary carcinogenesis in rats. Lipids. 1986;21:285–288. doi: 10.1007/BF02536414. [DOI] [PubMed] [Google Scholar]
  • 76.Reddy BS, Cohen LA, McCoy GD, Hill P, Weisburger JH, Wynder EL. Nutrition and its relationship to cancer. Adv Cancer Res. 1980;32:237–345. doi: 10.1016/s0065-230x(08)60363-2. [DOI] [PubMed] [Google Scholar]
  • 77.Jourdan ML, Maheo K, Barascu A, Goupille C, De Latour MP, Bougnoux P, Rio PG. Increased BRCA1 protein in mammary tumours of rats fed marine omega-3 fatty acids. Oncol Rep. 2007;17:713–719. [PubMed] [Google Scholar]
  • 78.Hardman WE, Avula CP, Fernandes G, Cameron IL. Three percent dietary fish oil concentrate increased efficacy of doxorubicin against MDA-MB 231 breast cancer xenografts. Clin Cancer Res. 2001;7:2041–2049. [PubMed] [Google Scholar]
  • 79.Shao Y, Pardini L, Pardini RS. Dietary menhaden oil enhances mitomycin C antitumor activity toward human mammary carcinoma MX-1. Lipids. 1995;30:1035–1045. doi: 10.1007/BF02536289. [DOI] [PubMed] [Google Scholar]
  • 80.Chen J, Hui E, Ip T, Thompson LU. Dietary flaxseed enhances the inhibitory effect of tamoxifen on the growth of estrogen-dependent human breast cancer (mcf-7) in nude mice. Clin Cancer Res. 2004;10:7703–7711. doi: 10.1158/1078-0432.CCR-04-1130. [DOI] [PubMed] [Google Scholar]
  • 81.Pardini RS. Nutritional intervention with omega-3 fatty acids enhances tumor response to anti-neoplastic agents. Chem Biol Interact. 2006;162:89–105. doi: 10.1016/j.cbi.2006.05.012. [DOI] [PubMed] [Google Scholar]
  • 82.Cannizzo F, Jr, Broitman SA. Postpromotional effects of dietary marine or safflower oils on large bowel or pulmonary implants of CT-26 in mice. Cancer Res. 1989;49:4289–4294. [PubMed] [Google Scholar]
  • 83.Iigo M, Nakagawa T, Ishikawa C, Iwahori Y, Asamoto M, Yazawa K, Araki E, Tsuda H. Inhibitory effects of docosahexaenoic acid on colon carcinoma 26 metastasis to the lung. Br J Cancer. 1997;75:650–655. doi: 10.1038/bjc.1997.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Paulsen JE, Elvsaas IK, Steffensen IL, Alexander J. A fish oil derived concentrate enriched in eicosapentaenoic and docosahexaenoic acid as ethyl ester suppresses the formation and growth of intestinal polyps in the Min mouse. Carcinogenesis. 1997;18:1905–1910. doi: 10.1093/carcin/18.10.1905. [DOI] [PubMed] [Google Scholar]
  • 85.Boudreau MD, Sohn KH, Rhee SH, Lee SW, Hunt JD, Hwang DH. Suppression of tumor cell growth both in nude mice and in culture by n-3 polyunsaturated fatty acids: mediation through cyclooxygenase-independent pathways. Cancer Res. 2001;61:1386–1391. [PubMed] [Google Scholar]
  • 86.Calviello G, Di Nicuolo F, Gragnoli S, Piccioni E, Serini S, Maggiano N, Tringali G, Navarra P, Ranelletti FO, Palozza P. n-3 PUFAs reduce VEGF expression in human colon cancer cells modulating the COX-2/PGE2 induced ERK-1 and -2 and HIF-1alpha induction pathway. Carcinogenesis. 2004;25:2303–2310. doi: 10.1093/carcin/bgh265. [DOI] [PubMed] [Google Scholar]
  • 87.Reddy BS, Maruyama H. Effect of dietary fish oil on azoxymethane-induced colon carcinogenesis in male F344 rats. Cancer Res. 1986;46:3367–3370. [PubMed] [Google Scholar]
  • 88.Minoura T, Takata T, Sakaguchi M, Takada H, Yamamura M, Hioki K, Yamamoto M. Effect of dietary eicosapentaenoic acid on azoxymethane-induced colon carcinogenesis in rats. Cancer Res. 1988;48:4790–4794. [PubMed] [Google Scholar]
  • 89.Takahashi M, Minamoto T, Yamashita N, Kato T, Yazawa K, Esumi H. Effect of docosahexaenoic acid on azoxymethane-induced colon carcinogenesis in rats. Cancer Lett. 1994;83:177–184. doi: 10.1016/0304-3835(94)90316-6. [DOI] [PubMed] [Google Scholar]
  • 90.Takahashi M, Fukutake M, Isoi T, Fukuda K, Sato H, Yazawa K, Sugimura T, Wakabayashi K. Suppression of azoxymethane-induced rat colon carcinoma development by a fish oil component, docosahexaenoic acid (DHA) Carcinogenesis. 1997;18:1337–1342. doi: 10.1093/carcin/18.7.1337. [DOI] [PubMed] [Google Scholar]
  • 91.Hendrickse CW, Keighley MR, Neoptolemos JP. Dietary omega-3 fats reduce proliferation and tumor yields at colorectal anastomosis in rats. Gastroenterology. 1995;109:431–439. doi: 10.1016/0016-5085(95)90330-5. [DOI] [PubMed] [Google Scholar]
  • 92.Iwamoto S, Senzaki H, Kiyozuka Y, Ogura E, Takada H, Hioki K, Tsubura A. Effects of fatty acids on liver metastasis of ACL-15 rat colon cancer cells. Nutr Cancer. 1998;31:143–150. doi: 10.1080/01635589809514694. [DOI] [PubMed] [Google Scholar]
  • 93.Kontogiannea M, Gupta A, Ntanios F, Graham T, Jones P, Meterissian S. omega-3 fatty acids decrease endothelial adhesion of human colorectal carcinoma cells. J Surg Res. 2000;92:201–205. doi: 10.1006/jsre.2000.5930. [DOI] [PubMed] [Google Scholar]
  • 94.Dwivedi C, Natarajan K, Matthees DP. Chemopreventive effects of dietary flaxseed oil on colon tumor development. Nutr Cancer. 2005;51:52–58. doi: 10.1207/s15327914nc5101_8. [DOI] [PubMed] [Google Scholar]
  • 95.Bommareddy A, Arasada BL, Mathees DP, Dwivedi C. Chemopreventive effects of dietary flaxseed on colon tumor development. Nutr Cancer. 2006;54:216–222. doi: 10.1207/s15327914nc5402_8. [DOI] [PubMed] [Google Scholar]
  • 96.Williams D, Verghese M, Walker LT, Boateng J, Shackelford L, Chawan CB. Flax seed oil and flax seed meal reduce the formation of aberrant crypt foci (ACF) in azoxymethane-induced colon cancer in Fisher 344 male rats. Food Chem Toxicol. 2007;45:153–159. doi: 10.1016/j.fct.2006.08.014. [DOI] [PubMed] [Google Scholar]
  • 97.Petrik MB, McEntee MF, Johnson BT, Obukowicz MG, Whelan J. Highly unsaturated (n-3) fatty acids, but not alpha-linolenic, conjugated linoleic or gamma-linolenic acids, reduce tumorigenesis in Apc(Min/+) mice. J Nutr. 2000;130:2434–2443. doi: 10.1093/jn/130.10.2434. [DOI] [PubMed] [Google Scholar]
  • 98.Newcomer LM, King IB, Wicklund KG, Stanford JL. The association of fatty acids with prostate cancer risk. Prostate. 2001;47:262–268. doi: 10.1002/pros.1070. [DOI] [PubMed] [Google Scholar]
  • 99.De Stefani E, Deneo-Pellegrini H, Boffetta P, Ronco A, Mendilaharsu M. Alpha-linolenic acid and risk of prostate cancer: a case-control study in Uruguay. Cancer Epidemiol Biomarkers Prev. 2000;9:335–338. [PubMed] [Google Scholar]
  • 100.Jatoi A, Rowland K, Loprinzi CL, Sloan JA, Dakhil SR, MacDonald N, Gagnon B, Novotny PJ, Mailliard JA, Bushey TI, et al. An eicosapentaenoic acid supplement versus megestrol acetate versus both for patients with cancer-associated wasting: a North Central Cancer Treatment Group and National Cancer Institute of Canada collaborative effort. J Clin Oncol. 2004;22:2469–2476. doi: 10.1200/JCO.2004.06.024. [DOI] [PubMed] [Google Scholar]
  • 101.Burns CP, Halabi S, Clamon G, Kaplan E, Hohl RJ, Atkins JN, Schwartz MA, Wagner BA, Paskett E. Phase II study of high-dose fish oil capsules for patients with cancer-related cachexia. Cancer. 2004;101:370–378. doi: 10.1002/cncr.20362. [DOI] [PubMed] [Google Scholar]
  • 102.Klek S, Kulig J, Szczepanik AM, Jedrys J, Kolodziejczyk P. The clinical value of parenteral immunonutrition in surgical patients. Acta Chir Belg. 2005;105:175–179. [PubMed] [Google Scholar]
  • 103.Harle L, Brown T, Laheru D, Dobs AS. Omega-3 fatty acids for the treatment of cancer cachexia: issues in designing clinical trials of dietary supplements. J Altern Complement Med. 2005;11:1039–1046. doi: 10.1089/acm.2005.11.1039. [DOI] [PubMed] [Google Scholar]
  • 104.Wigmore SJ, Barber MD, Ross JA, Tisdale MJ, Fearon KC. Effect of oral eicosapentaenoic acid on weight loss in patients with pancreatic cancer. Nutr Cancer. 2000;36:177–184. doi: 10.1207/S15327914NC3602_6. [DOI] [PubMed] [Google Scholar]
  • 105.Gogos CA, Ginopoulos P, Salsa B, Apostolidou E, Zoumbos NC, Kalfarentzos F. Dietary omega-3 polyunsaturated fatty acids plus vitamin E restore immunodeficiency and prolong survival for severely ill patients with generalized malignancy: a randomized control trial. Cancer. 1998;82:395–402. doi: 10.1002/(sici)1097-0142(19980115)82:2<403::aid-cncr21>3.0.co;2-1. [DOI] [PubMed] [Google Scholar]
  • 106.Burns CP, Halabi S, Clamon GH, Hars V, Wagner BA, Hohl RJ, Lester E, Kirshner JJ, Vinciguerra V, Paskett E. Phase I clinical study of fish oil fatty acid capsules for patients with cancer cachexia: cancer and leukemia group B study 9473. Clin Cancer Res. 1999;5:3942–3947. [PubMed] [Google Scholar]
  • 107.Mantovani G, Maccio A, Madeddu C, Gramignano G, Lusso MR, Serpe R, Massa E, Astara G, Deiana L. A phase II study with antioxidants, both in the diet and supplemented, pharmaconutritional support, progestagen, and anti-cyclooxygenase-2 showing efficacy and safety in patients with cancer-related anorexia/cachexia and oxidative stress. Cancer Epidemiol Biomarkers Prev. 2006;15:1030–1034. doi: 10.1158/1055-9965.EPI-05-0538. [DOI] [PubMed] [Google Scholar]
  • 108.Bruera E, Strasser F, Palmer JL, Willey J, Calder K, Amyotte G, Baracos V. Effect of fish oil on appetite and other symptoms in patients with advanced cancer and anorexia/cachexia: a double-blind, placebo-controlled study. J Clin Oncol. 2003;21:129–134. doi: 10.1200/JCO.2003.01.101. [DOI] [PubMed] [Google Scholar]
  • 109.Fearon KC, Von Meyenfeldt MF, Moses AG, Van Geenen R, Roy A, Gouma DJ, Giacosa A, Van Gossum A, Bauer J, Barber MD, et al. Effect of a protein and energy dense N-3 fatty acid enriched oral supplement on loss of weight and lean tissue in cancer cachexia: a randomised double blind trial. Gut. 2003;52:1479–1486. doi: 10.1136/gut.52.10.1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Dewey A, Baughan C, Dean T, Higgins B, Johnson I. Eicosapentaenoic acid (EPA, an omega-3 fatty acid from fish oils) for the treatment of cancer cachexia. Cochrane Database Syst Rev. 2007 doi: 10.1002/14651858.CD004597.pub2. CD004597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Cerchietti LC, Navigante AH, Castro MA. Effects of eicosapentaenoic and docosahexaenoic n-3 fatty acids from fish oil and preferential Cox-2 inhibition on systemic syndromes in patients with advanced lung cancer. Nutr Cancer. 2007;59:14–20. doi: 10.1080/01635580701365068. [DOI] [PubMed] [Google Scholar]
  • 112.Lai V, George J, Richey L, Kim HJ, Cannon T, Shores C, Couch M. Results of a pilot study of the effects of celecoxib on cancer cachexia in patients with cancer of the head, neck, and gastrointestinal tract. Head Neck. 2008;30:67–74. doi: 10.1002/hed.20662. [DOI] [PubMed] [Google Scholar]
  • 113.Nakamura K, Kariyazono H, Komokata T, Hamada N, Sakata R, Yamada K. Influence of preoperative administration of omega-3 fatty acid-enriched supplement on inflammatory and immune responses in patients undergoing major surgery for cancer. Nutrition. 2005;21:639–649. doi: 10.1016/j.nut.2005.03.001. [DOI] [PubMed] [Google Scholar]
  • 114.Zheng Y, Li F, Qi B, Luo B, Sun H, Liu S, Wu X. Application of perioperative immunonutrition for gastrointestinal surgery: a meta-analysis of randomized controlled trials. Asia Pac J Clin Nutr. 2007;16 Suppl 1:253–257. [PubMed] [Google Scholar]
  • 115.Aiko S, Yoshizumi Y, Tsuwano S, Shimanouchi M, Sugiura Y, Maehara T. The effects of immediate enteral feeding with a formula containing high levels of omega-3 fatty acids in patients after surgery for esophageal cancer. JPEN J Parenter Enteral Nutr. 2005;29:141–147. doi: 10.1177/0148607105029003141. [DOI] [PubMed] [Google Scholar]
  • 116.Heller AR, Rossel T, Gottschlich B, Tiebel O, Menschikowski M, Litz RJ, Zimmermann T, Koch T. Omega-3 fatty acids improve liver and pancreas function in postoperative cancer patients. Int J Cancer. 2004;111:611–616. doi: 10.1002/ijc.20291. [DOI] [PubMed] [Google Scholar]
  • 117.Bougnoux P, Hajjaju N, Baucher MA, Jordan ML, Goupille C, Body G, LeFoch O. Docosahexaenoic acid (DHA) intake during first line chemotherapy improves survival in metastatic breast cancer. Proc. Am. Assoc. Cancer Res. 2006;47:1237. (abstract #5276) [Google Scholar]
  • 118.Giardiello FM, Hamilton SR, Krush AJ, Piantadosi S, Hylind LM, Celano P, Booker SV, Robinson CR, Offerhaus GJ. Treatment of colonic and rectal adenomas with sulindac in familial adenomatous polyposis. N Engl J Med. 1993;328:1313–1316. doi: 10.1056/NEJM199305063281805. [DOI] [PubMed] [Google Scholar]
  • 119.Jacobs EJ, Rodriguez C, Mondul AM, Connell CJ, Henley SJ, Calle EE, Thun MJ. A large cohort study of aspirin and other nonsteroidal anti-inflammatory drugs and prostate cancer incidence. J Natl Cancer Inst. 2005;97:975–980. doi: 10.1093/jnci/dji173. [DOI] [PubMed] [Google Scholar]
  • 120.Solomon SD, McMurray JJ, Pfeffer MA, Wittes J, Fowler R, Finn P, Anderson WF, Zauber A, Hawk E, Bertagnolli M. Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N Engl J Med. 2005;352:1071–1080. doi: 10.1056/NEJMoa050405. [DOI] [PubMed] [Google Scholar]
  • 121.Bartoli GM, Palozza P, Marra G, Armelao F, Franceschelli P, Luberto C, Sgarlata E, Piccioni E, Anti M. n-3 PUFA and alpha-tocopherol control of tumor cell proliferation. Mol Aspects Med. 1993;14:247–252. doi: 10.1016/0098-2997(93)90011-2. [DOI] [PubMed] [Google Scholar]
  • 122.Bartram HP, Gostner A, Scheppach W, Reddy BS, Rao CV, Dusel G, Richter F, Richter A, Kasper H. Effects of fish oil on rectal cell proliferation, mucosal fatty acids, and prostaglandin E2 release in healthy subjects. Gastroenterology. 1993;105:1317–1322. doi: 10.1016/0016-5085(93)90135-y. [DOI] [PubMed] [Google Scholar]
  • 123.Bartram HP, Gostner A, Reddy BS, Rao CV, Scheppach W, Dusel G, Richter A, Richter F, Kasper H. Missing anti-proliferative effect of fish oil on rectal epithelium in healthy volunteers consuming a high-fat diet: potential role of the n-3:n-6 fatty acid ratio. Eur J Cancer Prev. 1995;4:231–237. doi: 10.1097/00008469-199506000-00004. [DOI] [PubMed] [Google Scholar]
  • 124.Aronson WJ, Glaspy JA, Reddy ST, Reese D, Heber D, Bagga D. Modulation of omega-3/omega-6 polyunsaturated ratios with dietary fish oils in men with prostate cancer. Urology. 2001;58:283–288. doi: 10.1016/s0090-4295(01)01116-5. [DOI] [PubMed] [Google Scholar]
  • 125.Demark-Wahnefried W, Robertson CN, Walther PJ, Polascik TJ, Paulson DF, Vollmer RT. Pilot study to explore effects of low-fat, flaxseed-supplemented diet on proliferation of benign prostatic epithelium and prostate-specific antigen. Urology. 2004;63:900–904. doi: 10.1016/j.urology.2003.12.010. [DOI] [PubMed] [Google Scholar]
  • 126.Rhodes LE, Shahbakhti H, Azurdia RM, Moison RM, Steenwinkel MJ, Homburg MI, Dean MP, McArdle F, Beijersbergen van Henegouwen GM, Epe B, et al. Effect of eicosapentaenoic acid, an omega-3 polyunsaturated fatty acid, on UVR-related cancer risk in humans. An assessment of early genotoxic markers. Carcinogenesis. 2003;24:919–925. doi: 10.1093/carcin/bgg038. [DOI] [PubMed] [Google Scholar]

RESOURCES